Download - Interferon-β induces loss of spherogenicity and overcomes … · 2014/2/13  · 1 Interferon-β induces loss of spherogenicity and overcomes therapy resistance of glioblastoma stem

Transcript
  • 1

    Interferon-β induces loss of spherogenicity and overcomes therapy resistance

    of glioblastoma stem cells

    Caroline Happold1, Patrick Roth1, Manuela Silginer1, Ana-Maria Florea4, Katrin

    Lamszus3, Karl Frei2, Rene Deenen4, Guido Reifenberger4,5, Michael Weller1

    1Laboratory of Molecular Neuro-Oncology, Department of Neurology, and

    2Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 26, and

    Neuroscience Center Zurich, 8091 Zurich, Switzerland; 3Laboratory for Brain Tumor

    Biology, Department of Neurosurgery, University Hospital Hamburg-Eppendorf,

    Martinistrasse 52, 20246 Hamburg, Germany; 4Department of Neuropathology (A.F.,

    G.R.) and Center for Biological and Medical Research (R.D.), Heinrich Heine

    University, Düsseldorf, Germany, 5German Cancer Consortium (DKTK), German

    Cancer Research Center (DKFZ), Heidelberg, Germany

    *Correspondence: Prof. Michael Weller, Department of Neurology, University

    Hospital Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland, Tel.: +41 (0)44

    255 5500, Fax: +41 (0)44 255 4507, E-mail: [email protected]

    Running title: Anti-glioma activity of interferon-β

    Keywords: glioma, interferon, MGMT, stem cell, temozolomide, irradiation

    This work was supported by a grant from the Swiss National Science Foundation

    (31003A_130122) to M.Weller.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 2

    Abbreviations

    CNPase, 2', 3'-cyclic nucleotide 3'-phosphodiesterase; DMSO, dimethylsulfoxide;

    EGF, epidermal growth factor; FGF, fibroblast growth factor; FCS, fetal calf serum;

    GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GFAP, glial fibrillary acidic

    protein; GIC, glioma-initiating cells; GO, gene ontology; IFN, interferon; IFNAR,

    interferon receptor; IL, interleukin; JAK, Janus kinase; LTL, long-term cell line; MFL,

    Mega-FAS-ligand; MGMT, O6-methylguanine DNA methyltransferase; MTT, 3-(4,5-

    dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PBS, phosphate-buffered

    saline; PI, propidium iodide; RMA, robust multi-array analysis; RT-PCR, reverse

    transcriptase PCR; SFI, specific fluorescence intensity; sh, small hairpin; si, small

    interfering; STAT, Signal Transducers and Activators of Transcription; STRING,

    Search Tool for the Retrieval of Interacting Genes/Proteins; STS, staurosporine;

    TMZ, temozolomide; TP53, tumor protein p53; TNFSF10/TRAIL, tumor necrosis

    factor-related apoptosis-inducing ligand; XAF-1, XIAP-associated factor 1; XIAP, X-

    linked inhibitor of apoptosis

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 3

    ABSTRACT

    Glioblastoma is the most common malignant brain tumor in adults and characterized

    by a poor prognosis. Glioma cells expressing O6-methylguanine DNA

    methyltransferase (MGMT) exhibit a higher level of resistance towards alkylating

    agents, including the standard of care chemotherapeutic agent, temozolomide (TMZ).

    Here, we demonstrate that long-term glioma cell lines (LTL) as well as glioma-

    initiating cell lines (GIC) express receptors for the immune modulatory cytokine

    interferon (IFN)-β and respond to IFN-β with induction of STAT-3 phosphorylation.

    Exposure to IFN-β induces a minor loss of viability, but strongly interferes with sphere

    formation in GIC cultures. Further, IFN-β sensitizes LTL and GIC to TMZ and

    irradiation. RNA interference confirmed that both IFN-β receptors, R1 and R2, are

    required for IFN-β-mediated sensitization, but that sensitization is independent of

    MGMT or TP53. Most GIC lines are highly TMZ-resistant mediated by MGMT

    expression, but nevertheless susceptible to IFN-β sensitization. Gene expression

    profiling following IFN-β treatment revealed strong up-regulation of IFN-β-associated

    genes including a proapoptotic gene cluster, but did not alter stemness-associated

    expression signatures. Caspase activity and inhibition studies revealed the

    proapoptotic genes to mediate glioma cell sensitization to exogenous death ligands

    by IFN-β, but not to TMZ or irradiation, indicating distinct pathways of death

    sensitization mediated by IFN-β. Thus, IFN-β is a potential adjunct to glioblastoma

    treatment that may target the GIC population. IFN-β operates independently of

    MGMT-mediated resistance, classical apoptosis-regulatory networks and stemness-

    associated gene clusters.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 4

    INTRODUCTION

    Glioblastomas are characterized by infiltrative growth and resistance to cell death

    induction. Despite multimodal therapy, tumor progression occurs inevitably, and

    survival remains in the range of months (1-3). Early therapy failure is associated with

    the expression of O6-methylguanine-DNA-methyltransferase (MGMT), a DNA repair

    protein that accounts for glioma cell resistance by counteracting the effects of

    alkylating chemotherapy (4). MGMT has become an important molecular marker and

    is now implemented in clinical diagnostics as predictive biomarker for benefit from

    alkylating chemotherapy and clinical outcome (5-7). Patients with a non-methylated

    MGMT promoter are prone to therapy failure with standard alkylating chemotherapy,

    and to date, effective approaches for this large group of patients, comprising more

    than half of all glioblastoma patients, are still lacking, including dose intensification of

    TMZ (8).

    Recently, a subfraction of glioma cells exhibiting stem cell-like properties (stem-like

    glioma cells), referred to as glioma-initiating cells (GIC), has been identified (9, 10).

    GIC are thought to have the ability of self-renewal, tumor initiation and pluripotency,

    and have been proposed to account for the ultimately lethal nature of glioblastoma.

    They may contribute to therapy resistance in vivo and therefore promote tumor

    progression. The value of GIC cultures and their MGMT status as a model to study

    resistance to TMZ in glioblastoma has remained controversial. Among a panel of 20

    GIC lines, some were sensitive to TMZ, which was associated with low MGMT

    protein levels, but not MGMT promoter methylation, and the MGMT promoter status

    was thus not strongly predictive of response to TMZ (11). High TMZ sensitivity of GIC

    cultures lacking MGMT expression has been described in vitro (12). Finally, the

    frequency of MGMT promoter-methylated alleles in glioblastomas may range from

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 5

    10% to 90%, but methylated alleles were enriched in GIC cultures (13). Considering

    the limited activity of current standards of care, new approaches should take into

    account this novel target cell population.

    IFN-β, a member of the interferon class I family, exerts numerous functions for

    cellular differentiation, cell growth and immune responses. IFN-β signaling is

    mediated through binding to a type II cytokine receptor, involving heterodimerization

    of 2 IFN-α/β receptor subunits, IFNAR-1 and IFNAR-2. Down-stream signalling

    pathways involve the Janus kinase/signal Transducers and Activators of

    Transcription (JAK/STAT) pathway (14) and lead to accumulation of MxA protein, a

    GTPase interfering with the cytoskeletal structure (15, 16). MxA induction is an

    established marker for responsiveness to IFN-α/β (17, 18).

    IFN-β has gained great clinical impact in the treatment of multiple sclerosis. Its safety

    profile in patients with brain disease has therefore been firmly established. Moreover,

    IFN-α/β were the first agents to show a significant survival benefit in randomized

    trials in malignant melanoma (19). The mechanisms mediating antitumor effects of

    IFN may involve direct cytostatic, anti-angiogenic or immune modulatory activities.

    IFN-β has been reported to sensitize human glioma cell lines to TMZ in a TP53- and

    MGMT-dependent manner (20, 21). These reports, however, were based on findings

    in T98G and U251MG, both being TP53 mutant cell lines (22, 23). Moreover, IFN-β

    has been attributed anti-stem cell properties in glioma models (24). Finally, clinical

    trials exploring the addition of IFN-β to radiotherapy plus TMZ have been initiated and

    interpreted as promising (25, 26). These data prompted us to evaluate the potential

    role of IFN-β as a component of future, innovative treatment approaches for

    glioblastoma in more detail. In brief, we demonstrate profound anti-GIC activity of

    IFN-β as well as IFN-β-mediated sensitization to TMZ and irradiation, but exclude

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 6

    MGMT and TP53 as primary mediators of these effects. IFN-β exposure induces

    characteristic IFN-β-associated gene clusters, but does not alter the stemness

    signature of GIC. A proapoptotic gene signature induced by IFN-β is functionally

    relevant, but mediates neither anti-GIC properties nor sensitivity to TMZ or

    radiotherapy.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 7

    MATERIALS AND METHODS

    Materials and cell lines

    The human U87MG and T98G glioma cell lines, K562 leukemia cells and MCF-7

    breast adenocarcinoma cells were purchased from the American Type Culture

    Collection (Rockville, MD, USA). The other long-term cell lines (LTL) were kindly

    provided by N. de Tribolet (Lausanne, Switzerland). LNT-229 cells cultured in our

    laboratory express TP53 wild-type transcriptional activity (23). The generation of

    LNT-229 cells depleted of TP or overexpressing MGMT has been described (23, 27).

    MGMT-silenced LN-18 cells were generated by transfection with pSUPER puro

    encoding MGMT short hairpin (sh) RNA using Metafectene Pro transfection reagent

    (Biontex Laboratories GmbH, Martinsried, Germany) (28). The GIC cell lines GS-2,

    GS-5, GS-7, GS-8 and GS-9 have been characterized elsewhere (29). LTL were

    cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal calf

    serum (FCS), 2 mM glutamine and penicillin (100 IU/ml) / streptomycin (100 μg/ml).

    GS sphere cultures were maintained in Neurobasal Medium® supplemented with 2%

    B-27 supplement, 1% GlutaMAX, 20 ng/ml epidermal growth factor, 20 ng/ml

    fibroblast growth factor and 32 IE/ml heparin. K-562 cells were maintained in RPMI-

    1640 supplemented with 1 mmol/L sodium pyruvate (Gibco Life Technologies) and

    10% fetal calf serum. TMZ was provided by Schering-Plough (Kenilworth, NJ, USA).

    A stock solution of TMZ at 200 mM was prepared in dimethylsulfoxide (DMSO).

    Recombinant IFN-β1b was purchased from AbD Serotec (Dusseldorf, Germany),

    reconstituted to a concentration of 1,000,000 IU/ml with distilled water. The following

    antibodies were used: STAT-3 and phospho-STAT-3, Caspase-3 and LC3A/B from

    Cell Signaling (Boston, MA, USA); glyceraldehyde 3-phosphate dehydrogenase

    (GAPDH) from Everest Biotech (Oxfordshire, UK); tumor necrosis factor-related

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 8

    apoptosis inducing ligand (TRAIL) and XIAP-associated factor 1 (XAF-1) from Santa

    Cruz (Santa Cruz, CA, USA); caspase 8 from Enzo Life Science (Framingdale, NY,

    USA); Glial fibrillary acidic protein (GFAP) from Dako (Carpinteria, CA, USA); Nestin

    from Zytomed Systems GmbH (Berlin, Germany); β-III tubulin from Abcam

    (Cambridge, UK). Mega-Fas-ligand (MFL) was provided by Topotarget (Copenhagen,

    Denmark). The MxA antibody was kindly provided by O. Haller, MD and G. Kochs,

    PhD (Department of Virology, University of Freiburg, Germany). All other reagents,

    including anti- 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) antibody, O6-

    benzylguanine (O6-BG) and salinomycin, were purchased from Sigma (St. Louis, MO,

    USA). For irradiation experiments, cells were irradiated in a Co-radiation source

    (Gebrüder Sulzer, Thermische Energiesysteme, 60-Co, Winterthur, Switzerland) at 1,

    3 and 5 Gy at 24 h after IFN-β exposure.

    Polymerase chain reaction

    Total RNA was prepared using the NucleoSpin System (Macherey-Nagel AG,

    Önsingen SO, Switzerland) and transcribed using random primers (Bioconcept/NEB,

    Bioconcept, Allschwil, Switzerland) and Superscript II reverse transcriptase

    (Invitrogen, Carlsbad, CA, USA). For reverse transcriptase PCR, the conditions were:

    35 cycles, 94°C/45 sec, specific annealing temperature (54°C for IFNAR-1, 52°C for

    IFNAR-2, 51°C for GAPDH) /45 sec, 72°C/1 min. For real-time (quantitative) RT-

    PCR, cDNA amplification was monitored using SYBRGreen chemistry on the 7300

    Real time PCR System (Applied Biosystems, Zug, Switzerland). The conditions were:

    40 cycles, 95°C/15 sec, 60°C/1 min. Data analysis was done using the ΔΔCT method

    for relative quantification. The following specific primers were used: GAPDH fwd: 5'-

    CTCTCTGCTCCTCCTGTTCGAC-3', GAPDH rv: 5'-TGAGCGATGTGGCTCGGCT-

    3'; IFNAR-1 fwd: 5'-TAT GCT GCG AAA GTC TTC TTG AG-3’; IFNAR-1 rv: 5'-TCT

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 9

    TGG CTA GTT TGG GAA CTG TA-3’; IFNAR-2 fwd: 5'-TCT TGA GGC AAG GTC

    TCG CTA-3’; IFNAR-2 rv: 5'-CAG GGA TGC ACG CTT GTA ATC-3’; XAF1 fwd: 5’-

    AGC AGG TTG GGT GTA CGA TG-3’; XAF1 rv: 5’-TGA GCT GCA TGT CCA GTT

    TG-3’; TRAIL fwd: 5’-TGC GTG CTG ATC GTG ATC TTC-3’ ; TRAIL rv: 5’-GCT CGT

    TGG TAA AGT ACA CGT A-3’.

    Flow cytometry, cell cycle and viability assays

    Differentiated glioma cells and sphere cultures were detached respectively

    dissociated using Accutase (PAA Laboratories, Pasching, Austria) and blocked with

    2% FCS in phosphate-buffered saline (PBS). The cells were incubated for 30 min on

    ice using the following PE-labeled antibodies: anti–IFNAR-1 or anti-IFNAR-2

    antibodies (PBL interferon source, Piscataway, NJ, USA) for IFNAR experiments and

    anti-CD133/2-PE (Miltenyi Biotec, Bergisch Gladbach, Germany) for stem cell marker

    experiments. Flow cytometry was performed with a CyAn® flow cytometer (Beckman

    Coulter, Nyon, Switzerland). Signal intensity was calculated as the ratio of the mean

    fluorescence of the specific antibody and the isotype control antibody (specific

    fluorescence index). Dead cells were gated out. For some analyses, cells were

    permeabilized by Fix/Perm Buffer Set (Lucerna Chem AG, Biolegend, Luzern,

    Switzerland). For analysis of cell death, cells were grown in six-well plates, incubated

    with IFN-β at 150 IU/ml for 24 h, washed in PBS and allowed to grow for 48 h.

    Annexin (Anx) V-fluorescein isothiocyanate (1:100) and propidium iodide (PI) (50

    µg/ml) were added, and fluorescence in a total of 10,000 events (cells) per condition

    was recorded in a CyAn flow cytometer. AnxV- or PI-positive cells were counted as

    dead cells, the remaining cells were designated the surviving cell fraction. In some

    experiments, loss of viability was also confirmed by trypan blue dye exclusion.

    Autophagic cell death was assessed by immunoblot and immunostaining. In brief, for

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 10

    the latter, the cells were exposed to IFN-β as indicated and Cytospin samples were

    prepared, fixed in 4% PFA, blocked in TBS containing 0.2% Triton X100, 5% goat

    serum and 5% horse serum, and exposed to the primary LC3A/B antibody (Cell

    Signaling, Boston, MA, USA) overnight at 4°C and 1 h at room temperature for

    secondary antibodies.

    Clonogenicity assays

    For LTL, clonogenicity assays were performed by seeding 100 cells (LNT-229, LN-

    18) per well in 96-well plates, allowed to adhere overnight, and exposed to IFN-β at

    150 IU/ml for 24 h in fresh medium. After removal of IFN-β, the cells were exposed to

    TMZ at the indicated concentrations for 24 h in serum-free medium, followed by an

    agent-free observation period for 7-14 days in serum-containing medium. Cell density

    was assessed by crystal violet staining. For sphere cultures, the cells were seeded at

    500 cells per well in neurobasal medium and treated consecutively as indicated

    above in neurobasal medium. Lower cell numbers resulted in inefficient sphere

    formation. Cell growth was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-

    diphenyltetrazolium bromide (MTT) assay. Initially, we confirmed that crystal violet

    assay and MTT assay were good surrogate markers of the number of colonies

    respectively spheres in these assays, but easier to standardize for large scale

    concentration response analyses.

    Immunoblot analysis

    The cells were treated as indicated and lysed in lysis buffer containing 50 mM Tris-

    HCl, 120 mM NaCl, 5 mM EDTA and 0.5% NP-40. Protein (20 µg/lane) was

    separated on 10% acrylamide gels. After transfer to nitrocellulose (Biorad, Munich,

    Germany), the blots were blocked in Tris-buffered saline containing 5% skim milk and

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 11

    0.05% Tween 20 and incubated overnight at 4°C with primary antibodies and 1 h at

    room temperature for secondary antibodies. Visualization of protein bands was

    accomplished using horseradish peroxidase-coupled secondary antibodies (Santa

    Cruz Healthcare, Santa Cruz, CA, USA) and enhanced chemiluminescence (Pierce /

    Thermo Fisher, Madison, WI, USA).

    Immunocytochemistry and immunofluorescence microscopy

    For stemness experiments, the cells were exposed to IFN-β (150 IU/ml) for 48 h and

    cytospin samples were prepared, fixed in 4% PFA and blocked in either blocking

    solution (Candor Bioscience GmbH, Wangen, Germany) for GFAP, β-III tubulin or

    CNPase, or PBS containing 10% swine serum and 0.3% Triton X for nestin, and

    exposed to the primary antibody overnight at 4°C and 30 min at room temperature for

    secondary antibodies. Detection was performed with DAB detection systems (Dako)

    for 5 min at room temperature, and slides were mounted in Eukitt (Sigma-Aldrich).

    Counterstaining was performed with hemalum. For IFNAR receptor staining, cytospin

    samples of LTL or GIC were prepared as described above. Staining was performed

    as suggested by the manufacturer, using IFNAR-1 antibody from antibodies-

    online.com (Atlanta, GA, USA) and IFNAR-2 antibody from Abcam (Cambridge, UK).

    RNA interference-mediated gene silencing

    For transient transfections, 2.5 × 105 glioma cells were seeded in a six-well plate and

    transfected with 100 nM of specific or scrambled control small interfering (si) RNA,

    using Metafectene Transfection reagent (Biontex). siRNA was purchased from

    Dharmacon / Thermo Fisher (Chicago, IL, USA) using siGENOME SMARTpool

    targeting human IFNAR-1 and human IFNAR-2. Samples were collected 48 h post

    transfection and processed for further treatment.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 12

    Reporter assay

    Dual luciferase/renilla assays were carried out with co-transfection of 150 ng of the

    specific reporter construct and 20 ng of the renilla reniformis-CMV (pRL-CMV) control

    plasmid (Promega, Madison, WI, USA). Luciferase activity was normalized to

    constitutive renilla activity. The pGL2-Luc MGMT construct (30) was a kind gift from

    Dr. S. Mitra (Sealy Center for Molecular Science and Department of Human

    Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston,

    TX, USA). The TP53-Luc construct has been described elsewhere (31).

    Caspase activity assay

    Cells were seeded at a density of 1000 cells/well in a six-well-plate and treated as

    described above with either IFN-β and TMZ alone or in combination with or without

    ZVAD-fmk, with either MFL (Mega-FAS-ligand) or staurosporine as positive control.

    Cells were incubated in lysis buffer (25 mM Tris/HCL, 60 mM NaCl, 2.5 mM EDTA,

    0.25% NP40) for 10 min, and the substrate Ac-DEVD-amc was added at a

    concentration of 20 μM. Fluorescence was assessed at 360 nm wavelength every 15

    min until extinction of fluorescence (32).

    Microarray-based gene expression profiling

    Technical details are provided in the supplementary methods. Array records are

    deposited under GEO accession number GSE53213.

    Data analysis

    Data are representative of experiments performed three times with similar results.

    Where indicated, analysis of significance was performed using the two-tailed

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 13

    Student’s t-test. Synergy of irradiation or TMZ and IFN-β was assessed by the

    fractional product method (33) where indicated and differences of 10% of observed

    versus predicted (additive) effect were considered synergistic. For the analysis of

    functional gene interactions, the Search Tool for the Retrieval of Interacting

    Genes/Proteins (STRING) Version 9.05 at http://string-db.org (34) was used. Highest

    confidence settings were applied, integrating combined scores higher than 0.900.

    Cluster analysis was performed by application of the Markov Cluster algorithm

    (MCL). Disconnected nodes were hidden from the image.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 14

    RESULTS

    IFNAR are expressed in human glioma cells

    In total, 12 LTL and 5 GIC were assessed for mRNA expression of IFNAR-1 and -2

    by RT-PCR. All lines expressed transcripts for both receptor subunits (Fig. 1A).

    IFNAR-1, in contrast to IFNAR-2, was not detected at the surface on any glioma cell

    line by flow cytometry (Fig. 1B, left panel), but was detected by immunostaining (Fig.

    1B, right panel). Permeabilization of the cells or scraping of cells instead of using

    accutase did not produce a specific signal for IFNAR-1 on flow cytometry either.

    Specific fluorescence intensity (SFI) values for IFNAR-2 above 3 were never seen in

    GIC, but in all LTL (data not shown).

    IFN-β signalling in glioma cells involves MxA and STAT-3

    To ensure that IFN-β induces classical signalling pathways in glioma cells, we

    investigated whether STAT-3, the proposed signal transducer in response to IFN-β in

    gliomas, and the IFN-α/β-regulated protein MxA, a classical marker for cellular

    responsiveness to IFN-β, were induced. Immunoblot confirmed that STAT-3

    phosphorylation levels increased shortly after exposure to IFN-β, with a peak at 30-

    60 min, with no relevant concentration dependency between 50 and 500 IU/ml, in

    LTL and GIC (Fig. 1C). Likewise, the exposure to IFN-β led to a concentration-

    dependent increase of MxA protein in LNT-229 (Fig. 1D, upper row) or LN-18 cells

    (data not shown), in a time-dependent manner (Fig. 1D, lower row), with first marked

    elevation of protein levels at a concentration of 150 IU/ml at 24 h.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 15

    IFN-β induces cell cycle arrest in glioma cells and causes sphere disruption in

    GIC

    Within the concentration and time ranges of our experiments, IFN-β induced a

    reduction of G1 cells in all cell lines, associated with increases in S cells or G2/M cells

    or both. Moreover, there was an increase in sub-G1 cells indicating minor cell death

    induction (Fig. 2A). A moderate induction of necrotic rather than apoptotic cell death

    was confirmed by AnxV/PI flow cytometry, defined by an increase of the PI-positive

    fraction (Fig. 2B). Cellular staining and immunoblot for autophagy were negative in

    response to IFN-β (Fig. 2C, D).

    We also assessed a possible modulation of spherogenicity of GIC by IFN-β. When

    exposed to 150 IU/ml IFN-β, singularized GIC cultures showed a reduced sphere

    formation at 1,000 – 10,000 cells per well; lower cell numbers did not result in

    efficient sphere formation, independent of IFN-β exposure (Fig. 2E). Conversely,

    exposure of fully formed spheres to 150 IU/ml IFN-β led to sphere disaggregation,

    with lesser and significantly smaller spheres remaining after 2 weeks of culture, as

    assessed by sphere count and absorbance (Fig. 2F). The single cells detaching from

    the sphere were viable during the first days after sphere disaggregation, but

    underwent cell death after a week, as assessed by trypan blue assay (data not

    shown).

    To assess the possible impact of IFN-β on stem cell differentiation, we analysed GS-

    2, 5, 7 and 9 cells for changes in the expression of different stem cell markers. GS-2

    and GS-9 cells expressed CD133, but expression remained unchanged after

    exposure to 150 IU/ml IFN-β for 48 h. GS-5 and GS-7 did not express CD133 (Fig.

    2G). Moreover, IFN-β-treated GS-2, GS-5 and GS-9 cells showed no changes of

    GFAP, nestin, β-III-Tubulin or CNPase staining (shown for GS-2) (Fig. 2H).

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 16

    Requirement for IFNAR-1 and IFNAR-2 for IFN-β-mediated sensitization to

    irradiation and TMZ

    To verify the role of IFNAR-1 and IFNAR-2 for IFN-β-mediated signal transduction

    and sensitization to TMZ and irradiation, we silenced the expression of both

    receptors by siRNA. Efficacy of gene silencing was assessed by RT-PCR for IFNAR-

    1, which could not be assessed by flow cytometry (Fig. 1), and by RT-PCR and flow

    cytometry for IFNAR-2 (Fig. 3A). In the setting of TMZ exposure (Fig. 3B) or

    irradiation (Fig. 3C), silencing of IFNAR-1 or -2 or both attenuated the sensitizing

    effect of IFN-β in LNT-229 cells and LN-18 cells, confirming a role for IFNAR-

    mediated signal transduction pathways in the IFN-β-induced sensitization process.

    IFNAR-2 gene silencing alone did not abrogate the effect of IFN-β on TMZ activity to

    the same extent as IFNAR-1 gene silencing alone.

    Response to TMZ, irradiation and IFN in GIC lines

    Since GIC are proposed to be the major source of tumor relapse and resistance, we

    next focused on these models. All investigated GIC, except GS-9, expressed MGMT

    and were highly resistant to TMZ (Supplementary Note 1, Fig. S1). IFN-β had no

    major effect on cell density of LTL cells, but a significant concentration-dependent

    impact of IFN-β alone was observed in GIC cells (Fig. 4A), as expected from the data

    shown in Fig. 2F. To assess whether IFN-β also partially overcomes TMZ resistance

    of GIC, we pre-exposed the cells to increasing concentrations of IFN-β (Fig. 4B, left

    panel), or to 150 IU/ml IFN-β for increasing periods of time (Fig. 4B, right panel), prior

    to treatment with TMZ at EC50 concentrations. All investigated GIC lines showed

    sensitization to TMZ after pre-exposure to IFN-β. Sensitization was significant in all

    cell lines when treated at a concentration of 150 IU/ml and for an exposure time of 24

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 17

    h. Exposure of GS-2 cells to IFN-β for 24 h before administration of TMZ induced a

    strong growth-inhibiting effect on the sphere cultures (Fig. 4C). IFN-β-treated cells

    showed a decrease in clonogenic survival exceeding 50% compared to TMZ only-

    treated cells, and exhibited a progressive loss of the sphere structure (see also Fig.

    2D). Similar effects, albeit with less impressive disaggregation, were observed for

    GS-5, GS-7 and GS-9 spheres, where cooperative effects were most prominent

    when IFN-β was given with TMZ at approximately EC50 concentrations.

    In the setting of irradiation experiments, GIC cells were not more resistant than LTL,

    with GS-2 displaying the strongest radioresistance of the panel, and GS-5 the

    weakest, possibly because of the TP53 wild-type status of this cell line (29) (Fig. 4D).

    Pre-exposure to increasing concentrations of IFN-β followed by low dose irradiation

    at 1 Gy resulted at least in additive inhibition of clonogenic survival. Again, in the

    rather radioresistant GS-2, a significant impact of IFN-β alone was observed (Fig. 4E,

    left panel); preexposure to IFN-β before irradiation led to a reduction of cell density

    fulfilling criteria of synergy, with an additional reduction of cell density of almost 50%

    when exposed to 150 IU/ml of IFN-β. GS-5 and GS-7 displayed synergistic effects at

    150 IU/ml IFN-β; no synergy was determined in GS-9 (Fig. 4E).

    Gene expression analyses

    Microarray-based gene expression profiling demonstrated IFN-β-dependent

    significant differential regulation of 509 genes after 6 h of IFN-β treatment in all three

    glioma cells lines investigated. Gene expression profiling after 24 h revealed

    significant differential regulation of 522 genes. The combined analysis, comparing the

    6 h and 24 h data, resulted in an overlap of 132 differentially expressed genes upon

    exposure to IFN-β in all three cell lines. In addition to the statistical analysis, where

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 18

    IFN-β treatment effects were studied globally across all three cell lines, the impact of

    IFN-β was assessed individually for every single cell line. Single analysis probe lists

    of all three cell lines were compared at 6 h and 24 h, respectively (Fig. S2), defining

    overlapping lists of regulated genes as well as lists of genes exclusively regulated in

    each individual cell line. Multiple transcripts known to be regulated by IFN-β were

    found, including STAT1, interferon regulatory factors (IRF) and Mx1 (MxA) (Table

    S1). Moreover, we noted that a number of genes predicted to promote apoptotic

    signaling were induced by IFN-β, (Table S1), with an overlap of 6 genes annotated to

    both gene groups. Submission of the differentially expressed IFN-regulated genes to

    the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) identified a

    single main cluster of genes with strong interaction profile (Fig. S3A). Submission of

    the differentially expressed apoptosis-related genes revealed 3 main clusters, one

    cluster involving tumor necrosis factor-related apoptosis-inducing ligand (TRAIL /

    TNFSF10)-interacting genes (yellow), one involving XIAP-associated factor 1 (XAF1)-

    interacting genes (blue), both connected via a third cluster centered around STAT1

    (green) (Fig. S3B). Induction of TRAIL and XAF1 was confirmed by RT-PCR and

    immunoblot analysis (Fig. 5). As we had observed differential reaction of the LTL cell

    line LNT-229 and the GIC cell lines GS-2 and GS-9 in clonogenic growth assays after

    exposure to IFN-β alone, we performed a gene ontology (GO) analysis of the genes

    exclusively regulated (FC > 2) in LNT-229, or the GIC lines (Fig. S2, peripheral

    regions), and identified several down-regulated genes involved in proliferation and

    cell growth, as well as up-regulated genes involved in negative regulation of

    proliferation or cell cycle arrest (Table S2). Notably, more genes were regulated in

    the GIC lines, with 2 main clusters of negative regulators of proliferation interacting

    mainly through JUN (blue cluster) and IL-8, IL-6 and JAK2 (red cluster) (Fig. S3C).

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 19

    Altered expression of apoptosis regulating genes does not mediate enhanced

    clonogenic cell death induced by TMZ or irradiation

    To define a role for altered expression of apoptotic machinery genes in the biological

    effects of IFN-β, we examined whether IFN-β alone or when combined with different

    death stimuli promoted caspase activation under the conditions that induced

    synergistic inhibition of clonogenic survival (Figure 4). As a positive control, we used

    MFL since we previously observed IFN-α-induced sensitization to CD95L although

    the mechanism remained unclear then (35). MFL, but neither IFN-β nor TMZ,

    induced the cleavage of caspases 8 or 3 or DEVD-amc-cleaving caspase activity

    (Fig. 6A, B, C). Similar results were obtained for irradiation assays (data not shown).

    Preexposure to IFN-β did not result in caspase processing in cells exposed to TMZ

    either. Cell death in the investigated settings was confirmed by trypan blue dye

    exclusion (Fig. 6D). Finally, we performed clonogenic cell death assays similar to

    those in Figure 4 in the absence or presence of the broad spectrum caspase

    inhibitor, zVAD-fmk, to explore whether caspase inhibition attenuated or abrogated

    the effects in clonogenic cell death assays of IFN-β or TMZ alone or in combination

    (Fig. 6E). These assays confirmed the absence of a role for caspases, either upon

    single agent exposure or combination. The biological activity of zVAD-fmk under

    these conditions was confirmed by the inhibition of death receptor-mediated

    apoptosis as previously reported (36).

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 20

    DISCUSSION

    Even after the implementation of TMZ as the first active chemotherapeutic agent

    when combined with standard radiotherapy, glioblastomas remain a major challenge

    in the field of neuro-oncology as they often relapse early and follow an invariably fatal

    clinical course. The latter is also true for the subgroup of patients with MGMT

    promoter methylation who derive most benefit from TMZ (4, 6). Large efforts have

    been made to improve prognosis by investigating new agents, either at recurrence,

    but recently more often early in development already in the paradigm of concomitant

    and adjuvant TMZ plus radiotherapy (3, 37, 38).

    Here we asked whether the immune modulatory cytokine, IFN-β, acts on glioma LTL

    and GIC lines, and sensitizes for the anti-clonogenic effects of TMZ or irradiation. We

    detected the expression of IFNAR-1 and -2 mRNA in all glioma cells, making them a

    potential target for IFN-β (Fig. 1A). Although detection of IFNAR-1 protein turned out

    to be challenging and was possible by immunofluorescence microscopy only, but not

    by flow cytometry, the biological effects of IFNAR-1 gene silencing were prominent:

    siRNA-mediated knockdown of the single IFNAR-1 chain inhibited IFN-β-mediated

    sensitization (Fig. 3), thereby confirming its biological function. Silencing of the

    IFNAR-2 chain alone inhibited IFN-β signalling only to a moderate level whereas

    silencing of IFNAR-1 was additionally required to abort the intracellular signal

    cascade enough to lead to a highly significant reduction in inhibition of clonogenic

    survival after IFN-β exposure (Fig. 3B,C). This may be due to the fact that, whilst

    IFNAR-2 holds only ligand-binding capacities, IFNAR-1, mainly inducing the

    intracellular signalling cascades, has also been described to have weak ligand

    binding activity, too, and may interact with other proteins in the absence of IFNAR-2

    (39, 40). Alternatively, although both siRNA pools reduced receptor mRNA

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 21

    expression to a similar extend (Fig. 3A), differential protein stability may account for

    different biological efficiency of gene silencing in these experiments. Responsiveness

    of the cells was assessed by monitoring the expression of target proteins of the IFN-β

    signaling pathways (Fig. 1D).

    STAT-3 has been shown to be involved in antiproliferative functions when induced by

    IFN-α/β in human Daudi cells (41) and, in glioma cells, may act as an IFN-β-induced

    tumor inhibitor through negative modulation of miR-21 (42). Conversely, there is also

    accumulating evidence that STAT-3 is a driver of the malignant phenotype of

    glioblastoma (43). In our study, STAT-3 was uniformly phosphorylated in both LTL

    and GIC cell lines shortly after exposure to IFN-β (Fig. 1C). Thus, the biological

    consequences of STAT-3 phosphorylation may be context-dependent and need to be

    interpreted in the natural course or the therapeutic setting in which they are

    observed.

    Since GIC cells cultured under sphere conditions are considered to be a more

    resistant subpopulation of glioma cells (44-46), we characterized cell cycle

    progression, cell death induction and sphere formation capacity of GIC in the

    presence of IFN-β. Similar to LTL, we observed a cell cycle arrest with a reduced G1

    phase and an increase of the sub-G1 population, reflecting a moderate increase in

    the necrotic fraction on AnxV/PI flow cytometry (Fig. 2A,B). Exposure of singularized

    cells isolated from sphere cultures to IFN-β led to reduced sphere formation;

    moreover, fully formed spheres exposed to IFN-β for 24 h disaggregated in the first

    days (Fig. 2E,F), and the cells underwent delayed cell death after more than a week

    as assessed by trypan blue dye exclusion (data not shown). This sphere disruption

    effect with delayed cell death induction appears to be a major effect of IFN-β on GIC

    survival and to play a more important role than early classical apoptotic pathways

    that were not shown to be activated in the first 48 h post exposure in AnxV/PI flow

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 22

    cytometry. Accordingly, Affymetrix array data demonstrated up-regulation of several

    apoptosis-related genes (Table S1, Fig. S3B), but ZVAD-fmk-controlled clonogenicity

    assays or DEVD-amc-cleaving-assays did again not confirm an induction of classical

    caspase-determined apoptotic pathways (Fig. 6C,E).

    We demonstrated that IFN-β facilitated the loss of clonogenicity induced by the

    standard chemotherapeutic agent for glioblastoma, TMZ. Since a sensitizing

    mechanism mediated through MGMT and TP53 had been described (20), we

    assessed IFN-β effects on a panel of cells with a heterogeneous pattern of MGMT

    and TP53 status (Supplementary Note 2, Fig.S4A). In our experimental settings,

    neither TP53 nor the MGMT status was important for the sensitizing effects of IFN-β.

    Specifically, we did not observe an induction of TP53 expression in cells with TP53

    wild-type transcriptional activity (LNT-229) or a down-regulation of MGMT expression

    in MGMT-expressing cells (LN-18) (Supplementary Note 2, Fig. S4C,D), nor did we

    observe MGMT- or TP53-associated changes in clonogenic survival assays, where

    sensitization by IFN-β was achieved independent of MGMT or TP53 status

    (Supplementary Note 2, Fig. S4A,B). Finally, Affymetrix chip analyses did neither

    detect induction of TP53 nor down-regulation of MGMT in the cell line expressing the

    respective gene products (TP53: LNT-229, GS-2; MGMT: GS-2; data not shown).

    The sensitization in the presence of MGMT is particularly attractive in view of the

    urgent clinical need for novel strategies for the majority of glioblastoma patients with

    tumors without MGMT promoter methylation. Moreover, we confirmed that

    preexposure to IFN-β sensitized different LTL to the anti-clonogenic effects of single

    doses of irradiation, too (Fig. S4B), and that these effects were mediated by the

    known receptors of IFN-β, IFNAR-1 and IFNAR-2 (Fig. 3).

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 23

    GIC lines, which were confirmed here to be highly resistant to TMZ due to MGMT

    expression, appeared to be a special target to IFN-β which impaired sphere formation

    when administered alone, in the absence of major cell death induction (Fig. 2).

    Accordingly, we observed a growth-inhibiting effect of IFN-β in GIC cultures even

    when applied as a single agent (Fig. 4A), even in MGMT-expressing and highly TMZ-

    resistant models (Fig. 4B-E). These chemosensitizing effects of IFN-β were

    concentration- and time-dependent, with significant results assessed at a

    concentration of 150 IU/ml, and a 24 h application (Fig. 4B, E). Of note, the molecular

    pattern in GIC cells was also diverse with regard to TP53 status (29), confirming that

    TP53 is not a major mediator of IFN-β-induced sensitization in glioma cells.

    All glioma models investigated here were sensitized at concentrations of IFN-β

    reached in the serum in clinical settings following intravenous application, where

    ranges of over 1000 IU/ml were reported at doses of 18x106 IE i.v. in healthy

    volunteers (47-49). IFN-β has been applied intravenously before in several clinical

    studies and was shown to be well tolerated without significant side effects (50),

    making it an interesting candidate for adjuvant glioblastoma therapy, close to

    practice. Thus, the present laboratory evidence, together with the wealth of data on

    the safety and tolerability of IFN-β in patients with multiple sclerosis, justifies further

    clinical trials of IFN-β in combination with radiotherapy and TMZ in patients with

    newly diagnosed glioblastoma, specifically those patients suffering from MGMT-

    unmethylated tumors.

    Acknowledgement

    The authors would like to thank Dr. Sankar Mitra and Dr. Kishor Bhakat for providing

    the MGMT reporter plasmid, as well as Jasmin Buchs, Nadine Lauinger and Silvia

    Dolski for excellent technical assistance.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 24

    Conflict of interest statement: C.H. has received honoraria for advisory board

    activity from MSD. P.R. has received honoraria for advisory board activity from MSD,

    Roche and Molecular Partners. M.W. received research support from Bayer,

    Antisense Pharma, Merck Serono and Roche and honoraria for advisory board and

    lecture activities from Merck Serono, MSD, Roche and Magforce. G.R. received

    honoraria for advisory board activities from Merck Serono and Roche. M.S., A. M. F.,

    K.L., R.D. and K.F. declare no conflict of interest.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 25

    References

    1. Johnson DR, O'Neill BP. Glioblastoma survival in the United States before and

    during the temozolomide era. J Neurooncol 2012;107:359-64.

    2. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et

    al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N

    Engl J Med 2005;352:987-96.

    3. Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med 2008;359:492-

    507.

    4. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al.

    MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med

    2005;352:997-1003.

    5. Weller M, Pfister SM, Wick W, Hegi ME, Reifenberger G, Stupp R. Molecular

    neuro-oncology in clinical practice: a new horizon. Lancet Oncol 2013;14:e370-9.

    6. Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W, et

    al. MGMT promoter methylation in malignant gliomas: ready for personalized

    medicine? Nat Rev Neurol 2010;6:39-51.

    7. Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, et al.

    Temozolomide chemotherapy alone versus radiotherapy alone for malignant

    astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol

    2012;13:707-15.

    8. Gilbert MR, Wang M, Aldape KD, Stupp R, Hegi ME, Jaeckle KA, et al. Dose-

    dense temozolomide for newly diagnosed glioblastoma: a randomized phase III

    clinical trial. J Clin Oncol 2013;31:4085-91.

    9. Park DM, Rich JN. Biology of glioma cancer stem cells. Mol Cells 2009;28:7-

    12.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 26

    10. Venere M, Fine HA, Dirks PB, Rich JN. Cancer stem cells in gliomas:

    identifying and understanding the apex cell in cancer's hierarchy. Glia 2011;59:1148-

    54.

    11. Blough MD, Westgate MR, Beauchamp D, Kelly JJ, Stechishin O, Ramirez AL,

    et al. Sensitivity to temozolomide in brain tumor initiating cells. Neuro Oncol

    2010;12:756-60.

    12. Beier D, Rohrl S, Pillai DR, Schwarz S, Kunz-Schughart LA, Leukel P, et al.

    Temozolomide preferentially depletes cancer stem cells in glioblastoma. Cancer Res

    2008;68:5706-15.

    13. Sciuscio D, Diserens AC, van Dommelen K, Martinet D, Jones G, Janzer RC,

    et al. Extent and patterns of MGMT promoter methylation in glioblastoma- and

    respective glioblastoma-derived spheres. Clin Cancer Res 2011;17:255-66.

    14. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells

    respond to interferons. Annu Rev Biochem 1998;67:227-64.

    15. Goetschy JF, Zeller H, Content J, Horisberger MA. Regulation of the

    interferon-inducible IFI-78K gene, the human equivalent of the murine Mx gene, by

    interferons, double-stranded RNA, certain cytokines, and viruses. J Virol

    1989;63:2616-22.

    16. Haller O, Kochs G. Interferon-induced mx proteins: dynamin-like GTPases with

    antiviral activity. Traffic 2002;3:710-7.

    17. Holzinger D, Jorns C, Stertz S, Boisson-Dupuis S, Thimme R, Weidmann M, et

    al. Induction of MxA gene expression by influenza A virus requires type I or type III

    interferon signaling. J Virol 2007;81:7776-85.

    18. von Wussow P, Jakschies D, Hochkeppel HK, Fibich C, Penner L, Deicher H.

    The human intracellular Mx-homologous protein is specifically induced by type I

    interferons. Eur J Immunol 1990;20:2015-9.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 27

    19. Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH.

    Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the

    Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 1996;14:7-17.

    20. Natsume A, Ishii D, Wakabayashi T, Tsuno T, Hatano H, Mizuno M, et al. IFN-

    beta down-regulates the expression of DNA repair gene MGMT and sensitizes

    resistant glioma cells to temozolomide. Cancer Res 2005;65:7573-9.

    21. Natsume A, Wakabayashi T, Ishii D, Maruta H, Fujii M, Shimato S, et al. A

    combination of IFN-beta and temozolomide in human glioma xenograft models:

    implication of p53-mediated MGMT downregulation. Cancer Chemother Pharmacol

    2008;61:653-9.

    22. Ishii N, Maier D, Merlo A, Tada M, Sawamura Y, Diserens AC, et al. Frequent

    co-alterations of TP53, p16/CDKN2A, p14ARF, PTEN tumor suppressor genes in

    human glioma cell lines. Brain Pathol 1999;9:469-79.

    23. Wischhusen J, Naumann U, Ohgaki H, Rastinejad F, Weller M. CP-31398, a

    novel p53-stabilizing agent, induces p53-dependent and p53-independent glioma cell

    death. Oncogene 2003;22:8233-45.

    24. Williams RF, Sims TL, Tracey L, Myers AL, Ng CY, Poppleton H, et al.

    Maturation of tumor vasculature by interferon-beta disrupts the vascular niche of

    glioma stem cells. Anticancer Res 2010;30:3301-8.

    25. Motomura K, Natsume A, Kishida Y, Higashi H, Kondo Y, Nakasu Y, et al.

    Benefits of interferon-beta and temozolomide combination therapy for newly

    diagnosed primary glioblastoma with the unmethylated MGMT promoter: A

    multicenter study. Cancer 2011;117:1721-30.

    26. Watanabe T, Katayama Y, Yoshino A, Fukaya C, Yamamoto T. Human

    interferon beta, nimustine hydrochloride, and radiation therapy in the treatment of

    newly diagnosed malignant astrocytomas. J Neurooncol 2005;72:57-62.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 28

    27. Hermisson M, Klumpp A, Wick W, Wischhusen J, Nagel G, Roos W, et al. O6-

    methylguanine DNA methyltransferase and p53 status predict temozolomide

    sensitivity in human malignant glioma cells. J Neurochem 2006;96:766-76.

    28. Maurer GD, Tritschler I, Adams B, Tabatabai G, Wick W, Stupp R, et al.

    Cilengitide modulates attachment and viability of human glioma cells, but not

    sensitivity to irradiation or temozolomide in vitro. Neuro Oncol 2009;11:747-56.

    29. Gunther HS, Schmidt NO, Phillips HS, Kemming D, Kharbanda S, Soriano R,

    et al. Glioblastoma-derived stem cell-enriched cultures form distinct subgroups

    according to molecular and phenotypic criteria. Oncogene 2008;27:2897-909.

    30. Biswas T, Ramana CV, Srinivasan G, Boldogh I, Hazra TK, Chen Z, et al.

    Activation of human O6-methylguanine-DNA methyltransferase gene by

    glucocorticoid hormone. Oncogene 1999;18:525-32.

    31. Wischhusen J, Melino G, Weller M. p53 and its family members -- reporter

    genes may not see the difference. Cell Death Differ 2004;11:1150-2.

    32. Wagenknecht B, Schulz JB, Gulbins E, Weller M. Crm-A, bcl-2 and NDGA

    inhibit CD95L-induced apoptosis of malignant glioma cells at the level of caspase 8

    processing. Cell Death Differ 1998;5:894-900.

    33. Webb JL. Enzyme and metabolic inhibitors. 1st ed New York: Academic press

    1963.

    34. Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, et al.

    STRING v9.1: protein-protein interaction networks, with increased coverage and

    integration. Nucleic acids research 2013;41:D808-15.

    35. Roth W, Wagenknecht B, Dichgans J, Weller M. Interferon-alpha enhances

    CD95L-induced apoptosis of human malignant glioma cells. J Neuroimmunol

    1998;87:121-9.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 29

    36. Glaser T, Wagenknecht B, Groscurth P, Krammer PH, Weller M. Death

    ligand/receptor-independent caspase activation mediates drug-induced cytotoxic cell

    death in human malignant glioma cells. Oncogene 1999;18:5044-53.

    37. Weller M, Stupp R, Hegi M, Wick W. Individualized targeted therapy for

    glioblastoma: fact or fiction? Cancer J 2012;18:40-4.

    38. Wick W, Weller M, Weiler M, Batchelor T, Yung AW, Platten M. Pathway

    inhibition: emerging molecular targets for treating glioblastoma. Neuro Oncol

    2011;13:566-79.

    39. Baychelier F, Nardeux PC, Cajean-Feroldi C, Ermonval M, Guymarho J, Tovey

    MG, et al. Involvement of the Gab2 scaffolding adapter in type I interferon signalling.

    Cell Signal 2007;19:2080-7.

    40. Cutrone EC, Langer JA. Contributions of cloned type I interferon receptor

    subunits to differential ligand binding. FEBS Lett 1997;404:197-202.

    41. Yang CH, Murti A, Pfeffer LM. STAT3 complements defects in an interferon-

    resistant cell line: evidence for an essential role for STAT3 in interferon signaling and

    biological activities. Proc Natl Acad Sci U S A 1998;95:5568-72.

    42. Ohno M, Natsume A, Kondo Y, Iwamizu H, Motomura K, Toda H, et al. The

    modulation of microRNAs by type I IFN through the activation of signal transducers

    and activators of transcription 3 in human glioma. Mol Cancer Res 2009;7:2022-30.

    43. Abou-Ghazal M, Yang DS, Qiao W, Reina-Ortiz C, Wei J, Kong LY, et al. The

    incidence, correlation with tumor-infiltrating inflammation, and prognosis of

    phosphorylated STAT3 expression in human gliomas. Clin Cancer Res

    2008;14:8228-35.

    44. Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, et al. Stem cell-

    like glioma cells promote tumor angiogenesis through vascular endothelial growth

    factor. Cancer Res 2006;66:7843-8.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 30

    45. Li Z, Wang H, Eyler CE, Hjelmeland AB, Rich JN. Turning cancer stem cells

    inside out: an exploration of glioma stem cell signaling pathways. J Biol Chem

    2009;284:16705-9.

    46. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al.

    Identification of human brain tumour initiating cells. Nature 2004;432:396-401.

    47. Buchwalder PA, Buclin T, Trinchard I, Munafo A, Biollaz J. Pharmacokinetics

    and pharmacodynamics of IFN-beta 1a in healthy volunteers. J Interferon Cytokine

    Res 2000;20:857-66.

    48. Chiang J, Gloff CA, Yoshizawa CN, Williams GJ. Pharmacokinetics of

    recombinant human interferon-beta ser in healthy volunteers and its effect on serum

    neopterin. Pharm Res 1993;10:567-72.

    49. Salmon P, Le Cotonnec JY, Galazka A, Abdul-Ahad A, Darragh A.

    Pharmacokinetics and pharmacodynamics of recombinant human interferon-beta in

    healthy male volunteers. J Interferon Cytokine Res 1996;16:759-64.

    50. Larocca AP, Leung SC, Marcus SG, Colby CB, Borden EC. Evaluation of

    neutralizing antibodies in patients treated with recombinant interferon-beta ser. J

    Interferon Res 1989;9 Suppl 1:S51-60.

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 31

    Figure legends

    Fig. 1. IFNAR-1 and -2 expression and responsiveness to IFN-β in human LTL

    and GIC. A,B. The cells were assessed for expression of IFNAR-1 and IFNAR-2

    mRNA by PCR with GAPDH as control (A) and of protein by cell surface flow

    cytometry (B, left panel; representative profiles for K562 as a positive control and

    LNT-229, LN-18, LN-308, GS-2 and GS-9 GIC cells; black curve: isotype control;

    grey curve: IFNAR antibody), or immunofluorescence staining (B, right panel; MCF-7

    and K562 were used as control). C. Phosphorylation levels of STAT-3 in a

    concentration- (left) and time-dependent (right) manner, shown representatively for

    LNT-229 cells (upper row), and for GS-7 and GS-9 GIC in a time-dependent manner

    (lower row). D. Responsiveness to IFN-β was assessed through detection of MxA

    protein in a concentration- (upper row) and time-dependent (lower row) manner,

    shown representatively for LNT-229 cells.

    Fig. 2. Assessment of IFN-β effects on GIC cells. A,B. LTL or GIC cells were

    exposed to IFN-β at 150 IU/ml for 24 h, cultured for an additional 48 h in serum-

    enriched medium, and flow cytometric cell cycle analysis (A) or AnxV/PI flow

    cytometry (B) were performed. Cell distributions are shown as bar graphs (striped:

    sub-G1, black: G1, white: S, grey: G2/M). Cell lines in B match those shown in A

    above. C. LNT-229 or GS-2 cells were exposed to IFN-β (150 IU/ml), ddH2O aqua

    (negative control) or salinomycin (SAL) (4 µM) (positive control) for 24 h. D. LNT-229

    and GS-2 cells were treated as in C for protein lysates and assessed for LC3 A/B

    cleavage. GAPDH was used as control. E. The effect of IFN-β (150 IU/ml, 24 h) on

    sphere formation over a range of 5 – 10,000 single cells was assessed by sphere

    count and is shown for GS-2 (black square, IFN-β; white diamond, vehicle). Data are

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 32

    expressed as mean ± SEM (n=6) (***p

  • 33

    effect of IFN-β is represented as delta of IFN-β-exposed versus vehicle-exposed

    cells.

    Fig. 4. GIC sensitization to TMZ and irradiation by IFN-β

    A. LTL or GIC cells were exposed to IFN-β in a concentration-dependent manner and

    clonogenic survival was assessed by crystal violet (LTL) or MTT (GIC) assay. B. LTL

    or GIC cells were exposed to IFN-β in a concentration-dependent (left) or time-

    dependent (right) manner before a 24 h pulse of TMZ in an EC50 concentration

    range. Clonogenic survival was assessed by crystal violet or MTT assay. C. GS-2,

    GS-5, GS-7 or GS-9 cells pre-exposed to IFN-β (150 IU/ml, 24 h) were treated with

    TMZ as shown in Fig. 3A and allowed to grow in neurobasal medium for 2 weeks

    before the assessment of clonogenicity by MTT assay (control: white bar; IFN-β:

    black bar). Data are expressed as mean ± SEM (*p

  • 34

    TRAIL were assessed by RT-PCR for RNA and by immunoblot for protein at 6 h and

    24 h after exposure to IFN-β in increasing doses (time points as assessed by micro-

    array profiling), in LNT-229 (left) and GS-2 (right).

    Fig. 6. Role of apoptosis-regulatory genes in IFN-β-induced sensitization.

    A, B. Immunoblots were performed after exposure of LNT-229 (left panel) or GS-2

    (right panel) to IFN-β for 6 h (upper row) or 24 h (lower row) and the levels of

    caspase 8 and 3 were assessed either for IFN-β exposure alone, TMZ alone, or in

    combination as described. MFL and STS were used as a positive control (MFL: 1

    μg/ml; STS: 1μM). C. DEVD-amc cleaving assays were performed after 6 h of

    exposure (shown for 2h fluorescence measurement). D. Cell death was assessed by

    trypan blue exclusion after 6 h of exposure to IFN-β at 150 or 300 IU/ml, IFN-β +

    TMZ (10 μM for LNT-229, 500 μM for GS-2), or MFL and STS as positive controls, as

    described in C; ddH2O was used as negative control (white bars, alive cells; black

    bars, dead cells; normalized to 100% cells). E. Clonogenic cell death assays were

    performed for LNT-229 and GS-2 and analyzed by crystal violet staining (for LNT-

    229) or MTT assay (for GS-2) at d10 after exposure to either IFN-β or TMZ alone, or

    combinations at indicated concentrations, with or without ZVAD-fmk (15 µM).

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • Fig. 1

    A

    IFNAR-1

    IFNAR-2

    GAPDHLN

    -18

    D24

    7MG

    U87

    MG

    LN-4

    28

    U13

    8MG

    T98

    G

    U25

    1MG

    LNT-

    229

    A17

    2

    LN-3

    19

    LN-3

    08

    U37

    3MG

    GS-7

    GS-2

    GS-5

    GS-9

    GS-8

    LTL GIC

    - 239 bp

    - 146 bp

    BIFNAR-1 IFNAR-2 IFNAR-1 IFNAR-2 Isotype

    K5

    62

    LN

    T-2

    29

    LN

    -18

    LN

    -30

    8G

    S-2

    GS

    -9

    MC

    F-7

    K5

    62

    LN

    T-2

    29

    LN

    -18

    GS

    -2G

    S-5

    C

    pSTAT-3

    STAT-3

    GAPDH

    0 50 150 300 500

    LNT-229

    pSTAT-3

    STAT-3

    GAPDH

    5 15 30 60 240

    GS-7

    IFN

    [IU/ml]

    min

    LNT-229

    5 15 30 60 240 min

    5 15 30 60 240 min

    GS-9

    D

    MxA

    GAPDH

    MxA

    GAPDH

    0 50 150 300 500 IFN

    [IU/ml]

    LNT-229

    LNT-229

    0’ 5’ 15’ 30’ 60’ 4h 24h 48h

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • 0%

    20%

    40%

    60%

    80%

    100%

    -IFN +IFN -IFN +IFN -IFN +IFN -IFN +IFN

    Fig. 2

    A

    B

    G2/M

    S

    G1

    Sub-G1

    CGS-2LNT-229

    ddH20

    IFN-β

    [150 IU/ml]

    SAL

    [µM]

    E

    LNT-229 LN-18 GS-2 GS-5

    F

    LC3 A/B-I

    LC3 A/B-II

    GAPDH

    LNT-229 GS-2

    IFN

    -

    [150IU

    /ml]

    SA

    L [4

    M]

    ddH

    2O

    IFN

    -

    [150IU

    /ml]

    SA

    L [4

    M]

    ddH

    2O

    D

    0

    20

    40

    60

    80

    1001

    Sp

    he

    res [n

    ] ***

    ******

    [n]

    1000 5000 10000

    Cells

    +IF

    N-β

    -IF

    N-β

    79.5

    31

    0

    20

    40

    60

    80

    100

    0

    20

    40

    60

    80

    100

    120

    ***

    ***

    untreated

    untreated

    IFN-β

    [150 IU/ml]

    IFN-β

    [150 IU/ml]

    IFN-

    + -

    G H1.8 1.5

    1 1

    1 1

    31 33

    + IFN-β [150 IU/ml]- IFN-β

    F1

    Sp

    he

    res [n

    ]

    10

    12

    A

    bso

    rba

    nce

    [%

    ]

    GS-2

    GS-5

    GS-7

    GS-9

    GFAP

    β -III-

    Tubulin

    CNPase

    Nestin

    + IFN-β [150 IU/ml]- IFN-β

    GS-2

    2.8% 3.1%

    93.8% 0.3%

    6.3%

    88.1%

    5.3%

    0.3%

    10.9% 5.3%

    77.3% 6.5%

    2.4%

    94.6% 1.5%

    1.5%

    19.8%

    77.9% 0.9%

    1.4%

    80.3% 1.4%

    13.8% 4.5%

    40.5%

    56.9%

    1.5%

    1.1%

    66.5% 1.1%

    31.2% 1.2%

    Annexin V Annexin VAnnexin VAnnexin V

    + IF

    N-β

    -

    IFN

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • Fig. 3

    A

    B

    C

    1

    0.8

    0.6

    0

    0.2

    0.4

    1

    0.8

    0.6

    0

    0.2

    0.4

    50

    40

    30

    0

    10

    20

    25

    20

    15

    0

    5

    10

    40

    30

    0

    10

    20

    60

    45

    0

    15

    30

    mR

    NA

    exp

    ressio

    n

    (no

    rma

    lize

    d to

    co

    ntr

    ol)

    Effe

    ct o

    f IF

    N [%

    ]E

    ffe

    ct o

    f IF

    N [%

    ]si_Ctrl si_IFNAR-2

    si_IFNAR-1 si_IFNAR-2

    LN

    T-2

    29

    LN

    -18

    LNT-229 LN-18

    LNT-229 LN-18

    **

    *

    *** ****** ***

    *********

    *********

    2.2

    1.32.4

    1.1

    knockdown

    si_Ctrl R2R1 R1+R2

    12.5 µM TMZ 500 µM TMZ

    5 Gy 5 Gy

    si_Ctrl R2R1 R1+R2

    si_Ctrl R2R1 R1+R2 si_Ctrl R2R1 R1+R2

    knockdown knockdown

    knockdown

    LNT-229 LN-18LN-18 LNT-229

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • Fig. 4

    A

    E

    D

    C

    B

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    20

    40

    60

    80

    0

    20

    40

    60

    80

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    120

    20

    40

    60

    80

    100

    0

    0 17 50 150 0 17 50 150 0 17 50 150 0 17 50 150 0 17 50 150

    LNT-229 GS-5GS-2LN-18 GS-9

    LNT-229 LN-18 GS-2 GS-5GS-9

    200 4000 6.25 12.5 25 50 100 800 0 62 125 250 500 1000

    0 2 4 6 8 100 62 125 250 500 1000

    GS-2 GS-5 GS-9GS-7

    GS-2 GS-5 GS-9GS-7GS-2 GS-5 GS-9GS-7

    Clo

    no

    ge

    nic

    ity [%

    ]C

    lon

    og

    en

    icity [%

    ]C

    lon

    og

    en

    icity [%

    ]C

    lon

    og

    en

    icity [%

    ]C

    lon

    og

    en

    icity [%

    ]C

    lon

    og

    en

    icity [%

    ]

    IFN-β

    [IU/ml]

    0

    1505017

    0

    - 24h- 4h- 1h

    1 Gy

    5 Gy3 Gy

    IFN-β

    [IU/ml]

    0

    1505017

    *****

    **

    ***

    ****** **

    **

    **

    ****

    ***

    ***

    ***

    **

    *

    * **

    *

    **

    *

    **

    **

    **

    ***

    ***

    ***

    ******

    ***

    ***

    ***

    ***

    ******

    ***

    *****

    **

    **

    *

    +

    +

    +

    +

    +

    1 GyCtrl

    GS-7 GS-9

    GS-5GS-2

    TMZ

    [µM]

    TMZ

    [µM]

    TMZ

    [µM]

    TMZ

    [µM]

    IFN-β [IU/ml]

    6.25 µM TMZ6.25 µM TMZ 500 µM TMZ 500 µM TMZ

    LNT-229 LN-18 GS-2 GS-5GS-9

    ***

    ******

    ***

    ***

    **

    **

    **

    ****

    **

    *

    **

    *

    *

    *

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • Fig. 5

    5000

    0

    1000

    2000

    3000

    4000

    800

    0

    200

    400

    600

    40

    0

    10

    20

    30

    20

    0

    4

    8

    12

    16

    30

    0

    10

    20

    600

    0

    200

    300

    400

    500

    100

    600

    0

    200

    400

    600

    0

    200

    300

    400

    500

    100

    XA

    F-1

    mR

    NA

    exp

    ressio

    n

    (no

    rma

    lize

    d to

    co

    ntr

    ol)

    TR

    AIL

    mR

    NA

    exp

    ressio

    n

    (no

    rma

    lize

    d to

    co

    ntr

    ol)

    LNT-229 GS-2

    6 h post IFN-β 24 h post IFN-β 6 h post IFN-β 24 h post IFN-β

    0 500300150500 50030015050 0 50030015050 0 50030015050

    0 500300150500 500300150500 500300150500 50030015050

    XAF-1

    GAPDH

    TRAIL

    0 50030015050 0 50030015050 0 500300150500 50030015050 IFN-β

    [IU/ml]

    IFN-β

    [IU/ml]

    IFN-β

    [IU/ml]

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/

  • Published OnlineFirst February 13, 2014.Mol Cancer Ther Caroline Happold, Patrick Roth, Manuela Silginer, et al. therapy resistance of glioblastoma stem cells

    induces loss of spherogenicity and overcomesβInterferon-

    Updated version

    10.1158/1535-7163.MCT-13-0772doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2014/02/19/1535-7163.MCT-13-0772.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://mct.aacrjournals.org/content/early/2014/02/13/1535-7163.MCT-13-0772To request permission to re-use all or part of this article, use this link

    on April 3, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on February 13, 2014; DOI: 10.1158/1535-7163.MCT-13-0772

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-13-0772http://mct.aacrjournals.org/content/suppl/2014/02/19/1535-7163.MCT-13-0772.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2014/02/13/1535-7163.MCT-13-0772http://mct.aacrjournals.org/

    Article FileFig.1Fig.2Fig.3Fig.4Fig.5Fig.6